Sie sind auf Seite 1von 9

REVIEW ARTICLE

published: 27 March 2014


doi: 10.3389/fphar.2014.00051

Mechanisms of cellular fibrosis associated with cancer


regimen-related toxicities
Maria L. Mancini* and Stephen T. Sonis
Biomodels LLC, Watertown, MA, USA

Edited by: Fibrosis is a common, persistent and potentially debilitating complication of chemotherapy
Lynne Anne Murray, MedImmune Ltd, and radiation regimens used for the treatment of cancer. The molecular mechanisms
UK
underlying fibrosis have been well studied and reveal overall processes that are largely
Reviewed by:
Darryl Knight, University of
ubiquitous. However, it is important to note that although the processes are similar,
Newcastle, Australia they result in cellular phenotypes that are highly tissue specific. These tissue specific
Lynne Anne Murray, MedImmune Ltd, differences may present opportunities for therapeutic interventions to prevent or treat this
UK often irreversible condition. Data generated from animal models of cancer therapy-related
*Correspondence: tissue toxicities have revealed that the signaling pathways involved in fibrosis are the same
Maria L. Mancini, Biomodels LLC, 313
Pleasant Street, Watertown,
as those involved in the normal injury response and include the transforming growth
MA 02472, USA factor β superfamily and a range of pro-inflammatory cytokines. The critical difference
e-mail: mmancini@biomodels.com between normal wound healing and fibrosis development appears to be, that in fibrosis,
these signaling pathways escape normal cellular regulation. As a result, an injury state
is maintained and processes involved in normal healing are usurped. There are a few,
if any, therapeutics that effectively prevent or treat fibrosis in patients. Consequently,
cancer survivors may be chronically plagued with a variety of life-altering fibrosis-related
symptoms. Uncovering the signaling pathways that drive cellular fibrosis is paramount
to the development of specific therapeutics that will mitigate this potentially devastating
condition.
Keywords: fibrosis, radiation therapy, chemotherapy toxicity, oral mucositis, radiation dermatitis, proctitis,
pulmonary fibrosis, TGF-ß

INTRODUCTION with cellular fibrosis of epithelial tissues and include radiation


Radiation and chemotherapy remain the most commonly used and/or chemotherapy-induced fibrosis of the gastrointestinal tract
therapies for the treatment of multiple types of human cancer. (oral mucositis and proctitis), the skin (dermatitis), and the lung
While these therapies have been met with great success in the treat- (pulmonary fibrosis).
ment of tumors, they are known to induce a wide range of acute
and chronic toxicities. These regimen-related toxicities are not MECHANISMS OF FIBROSIS
only associated with poor health outcomes, but they often become Within the past decade, there has been a major shift in the
dose-limiting for patients and impair patients’ quality of life (QoL) conventional paradigms associated with the pathogenesis of
and recovery in both the short and the long term (Elting et al., regimen-related toxicities in cancer patients. Historically, normal
2008). Hematological disorders such as anemia, thrombocytope- tissue damage was attributed to the concept that since radiation
nia, and neutropenia are among the most common complications or chemotherapy could not distinguish between rapidly divid-
associated with radiation and chemotherapy; however, cancer ing cancer cells and rapidly dividing normal cells the result was
patients are also at risk for a wide range of non-hematological tox- non-specific clonogenic cell death. Not only were the cellular
icities (Sonis and Keefe, 2013). The overall incidence of some form kinetics associated with normal tissue toxicity inconsistent with
of cancer treatment-related toxicity is almost 100% and can occur this hypothesis, but it failed to address damaging changes to
both during cancer treatment (acute toxicities), or will develop peripheral tissue such as subepithelial connective tissue or muscle
well after the completion of treatment (≥100 days, late toxicities). (i.e., heart) and completely ignored non-tissue based complica-
Understanding chemotherapy and radiation induced toxicities is tions like fatigue, cognitive dysfunction or cachexia. Accumulating
of high importance due to their direct impact on patients’ symp- evidence suggests that, while some clonogenic cell death does
toms and QoL and their high resource and financial burden. This occur, the bulk of pathogenesis is the consequence of a sequence of
review will focus on regimen-related toxicities that are associated related biological events that result in both direct and indirect tis-
sue and systemic damage mediated by a diverse range of canonical
Abbreviations: TGFβ, transforming growth factor β; QoL, quality of life; RT, radi- pathways. Of particular interest has been the finding that the tem-
ation therapy; ROS, reactive oxygen species; SOD, superoxide dismutase; CTGF,
poral genomic characterization of these toxicities has shown their
connective tissue growth factor; TNF-α, tumor necrosis factor alpha; INF-γ, inter-
feron gamma; EMT, epithelial to mesenchymal transition; Gy Gray, a unit of compatibility with conditions having similar phenotypes includ-
absorbed dose of ionizing radiation. ing chemotherapy-induced diarrhea and inflammatory bowel

www.frontiersin.org March 2014 | Volume 5 | Article 51 | 1


Mancini and Sonis Cancer regimen-induced fibrosis

disease (IBD), chemotherapy-fatigue and chronic fatigue syn- matrix (ECM) by the converting the flexible collagen type III into
drome, regimen-related cognitive dysfunction, and Alzheimer’s the more permanent collagen type I. This conversion is mediated
disease. While this observation has not been surprising, it does through both secreted proteases and matrix building proteins from
serve to emphasize the potential impact of better understanding local fibroblasts. Not surprisingly, heterogeneity exists among the
fibrosis in the context of cancer treatment, as well as possibly open- various fibroblast populations recruited; fibroblast subsets have
ing treatment intervention opportunities beyond the oncology specialized functions and vary in rates of proliferation, response
population. to inflammatory signals and ECM production (Sempowski et al.,
Data outlining molecular mechanisms by which treatment- 1995).
related fibrosis develops has largely been captured from animal Injury resulting from radiation and chemotherapy is initiated
models that accurately replicate chemotherapy or radiotherapy through two major paths: radiolytic hydrolysis and stimulation
regimens routinely used in patients (Sonis and Keefe, 2013). of the innate immune response. Of the two, oxidative stress is
Immediately following insult due to chemotherapy and radia- the best studied with respect to cancer treatment-associated tis-
tion, there is a large cellular response that involves cell type sue injury. Radiation or chemotherapy-induced oxidative stress
specific programs occurring in three general phases (Figure 1). leads to the production of oxygen free-radicals; specifically the
First is the inflammatory phase, where inflammatory cells are reactive oxygen species (ROS) superoxides, hydrogen peroxides,
recruited and release cytokines to recruit fibroblasts and other and hydroxyl radicals that cause oxidative damage to the tissue
immune cells to the site of injury. Second is the proliferative phase, (Pan et al., 2012). Once damage to the tissue has been initiated,
which is characterized by fibroblasts proliferating and migrating inflammatory cells are recruited to the injured area, a process
to the site of injury where they form a scaffold on a temporary orchestrated by vasodilation and vascular permeability. On the
fibronectin matrix present in the tissue and deposit collagen type cellular level, fibrosis involves the coordination of a variety of
III to form a new barrier. Third is the remodeling phase which cell types largely mediated through the fibroblast. The infiltrat-
lasts several weeks and involves building up the new extracellular ing immune cells secrete cytokines that drive the differentiation of

FIGURE 1 | Mechanisms of radiation and chemotherapy-induced fibrosis. Above is an illustration of the processes that drive fibrosis detailing the
mechanisms governing the overall tissue changes that occur as a result of radiation and chemotherapy induced injury; progressing from normal tissue to an
eventual fibrotic state.

Frontiers in Pharmacology | Inflammation Pharmacology March 2014 | Volume 5 | Article 51 | 2


Mancini and Sonis Cancer regimen-induced fibrosis

fibroblasts and other self-renewing cells into myofibroblasts which suggest that TGFβ/Smad signaling is directly involved in the induc-
deposit collagens and other ECM proteins at and around the site tion of fibrosis, other studies have shown that signaling through
of tissue damage (Eckes et al., 2000; Wang et al., 2011). Under nor- this pathway is not required for maintaining fibrosis and therefore
mal circumstances of wound repair, the expanded ECM would be therapies directed at regulating Smad activity would likely have
provisional until the process of re-epithelialization occurs. When to be administered as an early intervention (Holmes et al., 2001;
the wound repair process is deregulated, and re-epithelialization Leask et al., 2003). To further support this notion, the findings
is prevented, fibrosis occurs (Eckes et al., 2000; Ueha et al., 2012). of a recent meta-analysis of clinical data failed to demonstrate an
Until recently it was generally accepted that fibrosis occurs in a con- association between three SNPs commonly associated with TGFβ1
fined space only affecting the area immediately surrounding the and the risk of late radiation-induced normal tissue damage (Zhu
site of injury. It has since become clear that fibrosis arises and per- et al., 2013).
sists systemically. Circulating immune cells, chemokines, and bone CTGF is induced by TGFβ, specifically via Smad enhancer ele-
marrow derived fibroblasts are recruited to sites of injury gen- ments in the CTGF promoter, and therefore acts as a downstream
erating and depositing excess ECM proteins (Andersson-Sjöland effector of TGFβ mediated fibrosis (Leask et al., 2003). Data suggest
et al., 2011). On a molecular level, the processes of fibrosis are that after initial induction, expression of CTGF remains constitu-
driven by transforming growth factor beta (TGFβ), connective tis- tive in areas of fibrosis and escapes regulation by TGFβ signaling
sue growth factor (CTGF), tumor necrosis factor alpha (TNF-α), (Holmes et al., 2001). Therefore cellular events downstream of
and interferon gamma INF-γ (Reviewed in Leask and Abraham, CTGF secretion, including induction of collagen I expression in
2004). Susceptible organs include any connective tissue contain- neighboring cells, may contribute to the maintenance of fibrosis.
ing organ and/or tissues containing mesenchymal cells that are Furthermore, the paracrine nature of CTGF activity suggests a
capable of differentiating into fibroblasts. While the initial insult pivotal role in inducing fibrosis in surrounding tissues further
is often focused on a specific organ/tissue, systemic effects are seen exacerbating the duration and severity of fibrotic phenotypes
in distant sites highlighting the widespread nature of regimen- (Sonnylal et al., 2013). One mechanism of negative regulation
related toxicities. For example patients receiving radiation therapy of these signaling events involves the pro-inflammatory cytokine
(RT) with or without cytotoxics for head and neck cancer develop TNF-α which acts as an inhibitor of TGFβ induced CTGF expres-
diarrhea, supporting the idea that even directed therapies are sion and therefore has anti-fibrotic activity (Mori et al., 2002).
not contained to the treated tissues (Vermorken and Specenier, In addition, endogenous TNF-α applied directly to injured skin
2010). reduced the deposition of collagen. Some data suggests however,
The TGFβ signaling axis drives the majority of the cellular in certain situations sustained TNF-α expression may contribute
events associated with radiation-induced fibrosis (Boerma et al., to fibrosis in alternative mechanisms, through directly stimulat-
2013). The TGFβ superfamily regulates a wide variety of cellular ing the proliferation of fibroblasts (Piguet et al., 1990; Leask and
processes in response to injury including survival, proliferation, Abraham, 2004). That TNF-α does not regulate CTGF expression
and migration (Reviewed in Massague, 2012). TGFβ binds to its directly further supports the importance of CTGF related signaling
cognate receptors and channels its instructive signals through the events downstream of TGFβ in propagating fibrosis.
Smad family of transcription factors inducing the expression of Increased expression of the pro-inflammatory cytokine INF-γ
target genes involved in the cellular phenotypes described above goes hand in hand with tissue fibrosis. Naïve CD4+ T helper (Th)
and also upregulates the genes responsible for collagen synthesis cells differentiate into Th1 and Th2 subsets that secrete cytokines
(Ghosh et al., 2008). In addition, signaling through this path- in response to inflammation including INF-γ and a plethora of
way drives cellular dedifferentiation and reprogramming. Multiple interleukins essential for immune cell functions (Chen et al., 2001).
studies demonstrate that epithelial cells can undergo epithelial The specific role of INF-γ is incompletely understood; it is unclear
to mesenchymal transition (EMT), acquire the characteristics whether the presence of INF-γ at sites of fibrosis confers a role
of fibroblasts and subsequently differentiate into myofibroblasts. that is promoting or reparative. One study demonstrated that
This transition is driven by transcriptional changes that are medi- animals deficient in INF-γ did not develop pulmonary fibrosis
ated by TGFβ signaling and ultimately further exacerbate tissue resulting from treatment with Bleomycin suggesting a critical role
fibrosis (Venkov et al., 2007; Wang et al., 2011). Taken together, for this cytokine in promoting fibrosis (Chen et al., 2001). By con-
TGFβ acts both as a potent chemoattractant for fibroblasts and trast, other studies have shown that INF-γ has anti-fibrotic activity
subsequently a mediator of proliferation, migration, differentia- mediated through suppression of collagen synthesis by fibroblasts
tion, and ECM deposition of the recruited fibroblasts at the site of and inhibition of TGFβ expression (Gurujeyalakshmi and Giri,
injury. 1995). Therefore, data suggest that spatiotemporal expression of
The molecular mechanisms of TGFβ signaling associated with INF-γ may determine the role it plays in the development and
the development of fibrosis have been underscored through genet- maintenance of fibrosis. This is similar to the dichotomous role
ically modified animal models (Chen et al., 2001; Flanders et al., for TNF-α, the pro-inflammatory cytokine that is also regulated by
2002; Chan et al., 2012; Balli et al., 2013). Epithelium in animals and synergizes with INF-γ (Nathan et al., 1984). Taken together the
engineered to suppress Smad3 has a reduced fibrotic response signaling events regulating fibrosis represent distinct yet interde-
(Flanders et al., 2002; Oh et al., 2012). Furthermore, Smad3 pendent signaling pathways that become de-regulated in situations
null animals demonstrate accelerated wound repair characterized of radiation and chemotherapy-induced injury. These pathways
by enhanced re-epithelialization with a reduced inflammatory provide options for therapeutic interventions (Table 1) and will
response (Ashcroft et al., 1999). While the data from these studies be categorically explained in greater detail.

www.frontiersin.org March 2014 | Volume 5 | Article 51 | 3


Mancini and Sonis Cancer regimen-induced fibrosis

Table 1 | Current standard of care therapies for common regimen-related toxicities.

Regimen-related toxicity Treatment Mechanism

Oral mucositis MuGuard, gelclair Promotes barrier function


Caphosol, kepivance Topical biologics designed to promote healing

Proctitis 5-ASA, steroids Anti-inflammatory


Sucralfate, metronidazole Promotes barrier function, prevents bacterial colonization

Dermatitis Aloe vera Largely unknown; limited barrier function and lubrication

Pulmonary fibrosis Amifostine Produces cytoprotective thiols


Superoxide dismutases (SODs) Dismutate superoxides; prevents oxidative damage

Examples of therapies used to treat patients suffering with the toxicities described in this article. None of these therapies are specifically targeted to address fibrosis
directly; rather they are designed to prevent the initial inflammatory responses that eventually lead to fibrosis for purposes of prevention. This strategy has been met
with limited success and therefore there is an unmet clinical need for development of therapeutics focused specifically on fibrosis.

FIBROSIS ASSOCIATED WITH ORAL MUCOSITIS largely occurs in the endothelium and connective tissue of the
Radiotherapy is a treatment mainstay for cancers of the head and submucosa after exposure to radiation (Sonis, 2007). In response
neck and is typically administered concomitantly with radiosen- to the activation of a range of canonical pathways, a cytokine cas-
sitizing doses of chemotherapy, most commonly Cisplatin (Sonis, cade follows as does simultaneous fibronectin breakdown, and
2011). The oral, oropharyngeal, hypopharyngeal, and laryngeal amplification of pro-inflammatory cytokine signaling cascades in
mucosa are often included in the radiation field. As a consequence the aforementioned TGFβ and TNF-α pathways. Ultimately the
severe tissue injury in the form of mucositis is virtually ubiquitous. epithelium breakdowns down and ulceration occurs (Sonis et al.,
Lesions of oral mucositis consist of diffuse, deep, extremely painful 2000). Secondary bacterial colonization and breaks in the mucosal
ulcers involving the mouth’s movable mucosa (Sonis, 2013). As in barrier allow for penetration of whole bacteria or, more com-
most treatment paradigms, radiation is in small, daily fractionated monly, cell wall products which activate infiltrating macrophages
doses of 2Gy, 5 days per week, for cumulative doses of 60–70Gy to produce additional cytokines. The injured mucosa undergoes
(Silverman, 2007). Chemotherapy is given either every 3 weeks extensive remodeling to seal off the tissue to try and prevent inva-
during the radiation period (days 0, 21, and 42) or weekly, but sion of the bacteria. This process involves cellular deposits of ECM
in smaller doses. These treatment schemes induce a predictable that, as with many situations of wound repair, can encompass the
pattern of mucositis. By the end of the first week of treatment surrounding tissue and result in extensive fibrosis.
(cumulative radiation dose of 10Gy) erythematous changes start Clinically, patients with fibrosis of the mouth develop trismus
to occur and are accompanied by a level of pain described as being and cannot function normally (Lyons et al., 2013). But fibrosis
comparable to a bad food burn. By cumulative doses of 30Gy, is not limited to the mouth. Salivary function is obscured by
ulceration develops (Silverman, 2007; Sonis, 2013). Unlike the replacement of parenchyma with fibrous tissue and esophageal
typical mouth sores of aphthous stomatitis (canker sores), ulcer- strictures can develop (Fujita-Yoshigaki and Qi, 2009). While
ative lesions associated with mucositis are more broad and deep. mucositis is an acute toxicity, fibrosis-related changes tend to
Consequently, they are disproportionately painful requiring opi- be more chronic and are thus increasingly significant as cancer
oid analgesics which are often ineffective. Patients frequently are survivorship improves. Animal models of mucositis in rats, mice
unable to eat by mouth and require gastrostomy tube placement and hamsters have provided highly useful templates to define the
for feeding (Sonis, 2007). Subepithelial changes that accompany pathogenesis of mucosal injury and evaluate the potential effi-
mucositis predispose patients to fibrosis and the clinical devel- cacy of new interventions. Among the models routinely used, a
opment of trismus, which is characterized by a restricted ability highly translatable model in hamsters has been especially useful in
to open the mouth sometimes referred to as “lock jaw” (Lyons drug development and in demonstrating the relationship between
et al., 2013). Of regimen-related tissue injuries, the pathogenesis radiation-induced mucosal damage and the development of fibro-
of mucositis is probably best understood and has been described sis (Ara et al., 2008; Murray et al., 2010). An approved, effective
as a 5-phase algorithm which it appears to share with other types mechanistically based therapy for radiation-induced mucositis is
of cancer regimen-induced epithelial damage (Sonis et al., 2000). being aggressively sought. It is likely that a halo benefit from such
While originally directed at the oral mucosa, it is now clear that a treatment will be attenuation of fibrosis development.
the biological sequence is the same for regimen-related damage
throughout the gastrointestinal tract. FIBROSIS ASSOCIATED WITH PROCTITIS
As noted above, the initiation phase is characterized by oxida- Radiation-induced proctitis is a complication resulting from
tive stress and activation of the innate immune response which radiation directed at the lower abdomen or pelvis typically

Frontiers in Pharmacology | Inflammation Pharmacology March 2014 | Volume 5 | Article 51 | 4


Mancini and Sonis Cancer regimen-induced fibrosis

associated with rectal, prostate, or cervical malignancies. The any, compelling clinical studies in the treatment of radiation proc-
incidence of proctitis is approximately 75% with symptoms titis, the majority of therapies are based on the results of small
consisting of rectal bleeding, mucus production and diarrhea unblinded studies with mixed results. Animal models that utilize
(Counter et al., 1999). Not surprisingly given the nature of rec- focused radiation to the bowel have been effective tools for screen-
tal epithelium, symptoms begin within 2–3 weeks after the start ing potential compounds because the inflammatory responses that
of treatment. In contrast to lesions of the upper gastrointestinal occur in these models accurately represent the diversity seen in
tract, however, proctitis often lasts anywhere from a few months patients (Skwarchuk and Travis, 1998; Kang et al., 2000; Symon
to several years following the completion of radiation therapy. et al., 2010). The most common first-line therapies have been
The stages are multifactorial beginning with initial damage to adapted from the treatments used in IBD, including the anti-
the mucosa, followed by delayed slow growing connective tis- inflammatory compounds 5-ASA and steroids, sucralfate which
sue and finally a tissue response to vascular ischemia (Okunieff promotes barrier function and metronidazole to mitigate bacterial
et al., 2005). In addition to rectal bleeding and mucus, patients colony formation (Symon et al., 2010). Increasingly, endoscopic
suffering from proctitis also experience tenesmus, or a feeling therapies are being employed to control bleeding which include
or inability to empty the bowel upon defecation. This occurs heat probes, lasers, and most commonly argon plasma coagu-
as a result of epithelial fibrosis in the rectum due to radiation lation (APC). APC involves the flow ionized argon gas and can
exposure, a condition which is often permanent and irreversible target small or larger areas of bleeding without making physical
(Symon et al., 2010). contact to the tissue (Leiper and Morris, 2007). Other studies
The overall pathogenesis of fibrosis associated with radiation- suggest that long term suppression of high pro-inflammatory
induced proctitis is similar to that which occurs in oral mucositis. cytokine levels while also stimulating with pro-survival growth
However, unlike oral mucositis, radiation-induced proctitis has factors will provide the right balance to prevent long term com-
not been as aggressively studied pre-clinically and there exists a plications (Okunieff et al., 2005). Linard et al. (2013) recently
very high unmet clinical need in this area. Proctitis is initiated reported a different therapeutic approach focusing on prevent-
on the cellular level with apoptosis, disruptions of mitosis, and ing the progression of fibrosis with autologous mesenchymal stem
fibroblastic proliferation that leads to swelling and sloughing of cells (MSCs). Using an irradiated pig model, they initiated infu-
the rectal mucosa (Haboubi et al., 1988; Brunn and Fletcher, 2006). sion of MSCs following endoscopic identification of early fibrosis.
It has become increasingly clear that the complications associ- MSCs limited the progression of radiation-induced fibrosis due to
ated with proctitis involve coordination between the processes a reduction in collagen deposition, a decreased transforming TGFβ
of fibrosis and angiogenesis. The cellular alterations to the vas- response and modification of matrix metalloproteinase/TIMP bal-
culature including neovascularization and telangiectasias lead to ance. The novelty of this tactic provided a new arena for developing
clinical symptoms of persistent bleeding. Increased fibrosis causes potential therapeutic strategies that may address these currently
ischemia and eventual necrosis of the bowel tissue (Haselton et al., irreversible complications.
1985; Fajardo, 2005). The exact mechanisms for the late changes
in vascularity and fibrosis have yet to be elucidated, however, there FIBROSIS ASSOCIATED WITH RADIATION DERMATITIS
is evidence that several growth factors including platelet-derived Fibrosis of the skin in the context of cancer treatment occurs
growth factor, vascular endothelial growth factor, and fibroblast as a result of radiation-induced dermatitis. Radiation dermatitis
growth factor play key roles in the pathology (Brunn and Fletcher, is a common side effect of radiation therapy. It has a reported
2006) More recent evidence has suggested a role for mast cell frequency of 85% and is commonly seen in patients receiving
involvement based on the synergistic expression of endothelial and radiotherapy for the treatment of the breast, lung, prostate or
inflammatory genes in response to radiation, including p38a MAP head and neck cancers. The fibrosis which often accompanies
kinase and p65 (NF-κB; Blirando et al., 2011). Given the findings dermatological changes, results in symptoms similar to contrac-
that serine peptidases, particularly tryptases, secreted by mast tures that markedly compromise patients’ ability to move freely.
cells are able to stimulate both fibroblast chemotaxis and collagen Examples include patients treated for breast cancer have a lim-
production, the potential significance of mast cells in the patho- ited range of motion of their arm, or head and neck cancer
genesis of fibrosis definitely warrants additional investigation patients who have hindered head movement. The clinical presenta-
(Hugh and Pemberton, 2002; Caughey, 2007). Furthermore, radi- tion varies from mild erythema to complete epithelial breakdown
ation induced an increase in expression of αv β3 integrin (Abdollani manifested by painful ulceration and fibrosis (Ryan, 2012). Injury
et al., 2005). The αv β3 is highly expressed on endothelial cells and to the skin occurs immediately following the first dose of radi-
is known to have potent angiogenic activity involving prolifera- ation, disrupting the self-renewing cells of the epidermis (Ryan,
tion and migration of endothelial cells mediated through FGF and 2012). Subsequent doses prevent this cell population from fully
therefore likely contributes to the vascular changes observed in replenishing itself and also elicit an inflammatory response result-
radiation proctitis (Son et al., 2013). Taken together, these mech- ing in systemic complications. Later doses further exacerbate the
anisms may individually or collectively contribute to long term alterations in cellular phenotypes and can cause chronic issues
defects in tissue and vascular integrity associated with propagat- including delayed ulcerations, fibrosis and in the most severe cases,
ing the injury response mechanisms that promote fibrosis (Sheth necrosis of the tissue (Bey et al., 2010; Salvo et al., 2010).
et al., 2009). After an initial exposure to radiation, there is immediate dam-
Current treatment options for radiation proctitis vary greatly age to the keratinocyte cells of the skin, which is accompanied
in success rates (Leiper and Morris, 2007). While there are few, if by a simultaneous increase in free-radicals, DNA damage and

www.frontiersin.org March 2014 | Volume 5 | Article 51 | 5


Mancini and Sonis Cancer regimen-induced fibrosis

inflammation (Brown and Rzucidlo, 2011; Ryan, 2012). Like other the radiated fields include the thorax which exposes the lungs to
epithelial tissues, many of these biological changes are apparent in the potential for injury. Radiation directed to the thoracic region
the dermis. The TGFβ signaling pathway is largely implicated in exceeding 50Gy damages pulmonary tissues and may lead to the
driving the pathogenesis of radiation-induced dermatitis where development of dose-limiting pneumonitis or fibrosis (Reviewed
TGFβ levels are markedly increased in irradiated skin and remain in Tsoutsou and Koukourakis, 2006). Moderate to severe radiation
elevated for a long period of time (Martin et al., 2000; Flanders pneumonitis occurs with a reported incidence of between 10 and
et al., 2003). TGFβ isoforms act as potent chemotactic factors for 20% (Mehta, 2005) and is associated with alveolar damage that
monocytes, neutrophils, mast cells, and fibroblasts. Furthermore, causes a significant inflammatory response in the interstitial space
the Smad family of proteins regulates expression of additional of the lung. The increased vascular permeability causes edema
genes responsible for inflammation and fibrosis amplifying and and accumulation of proteins in the alveolar space (Rosai, 1996).
maintaining the cascade of inflammatory signals (Flanders et al., Pneumonitis develops within 12 weeks of receiving radiation ther-
2002). Smad3 null mice exposed to ionizing radiation exhibited apy and symptomatically manifests by shortness of breath. Severe
decreased skin damage and reduced fibrosis compared to wild cases increase the risk of treatment-related death significantly.
type animals, further supporting a critical role for this signaling Ultimately, pneumonitis may transition to pulmonary fibrosis.
pathway in the phenotypes associated with radiation dermatitis Pulmonary fibrosis is among the most thoroughly studied
(Flanders et al., 2002, 2003). cancer regimen-induced forms of fibrosis. Although it can be
There is currently no effective intervention to prevent or favor- caused by a variety of chemotherapeutic drugs or by radiation
ably modify the course and severity of radiation dermatitis (Wong and is associated with poor clinical outcomes, its pathogenesis
et al., 2013). Treatment strategies have been based on those associ- is incompletely understood. Of chemotherapies associated with
ated with thermal burns. Consequently, there is an unmet clinical pulmonary fibrosis, cases resulting from bleomycin treatment are
need to develop specific therapies to target the mechanisms under- the best studied. Radiation-induced fibrosis has been investigated
lying the development and persistence of radiation dermatitis in parallel and emerging data suggests the biological pathways
(Lataillade et al., 2007; Bey et al., 2010; Salvo et al., 2010; Chan which impact end organ damage are likely to be similar. Radiation-
et al., 2012). Pre-clinical animal models of radiation-induced der- induced pulmonary fibrosis is mediated through inflammatory
matitis in the published literature are limited however, a few cells that are recruited and accumulate in the interstitial space.
recent studies performed in mouse models have shown promis- It is believed that these cells are responsible for the majority of
ing therapeutic efficacy signals. In one example, treatment with a TGFβ production which drives the differentiation of fibroblasts to
Toll-like receptor 5 agonist reduced radiation dermatitis associated myofibroblasts depositing collagens into the ECM (Eckes et al.,
epidermal hyperplasia and dermal inflammation through activa- 2000; Wang et al., 2011). In addition, the pulmonary alveolar
tion of endogenous antioxidants reducing free-radicals (Burdelya type II epithelial cells can undergo EMT and differentiate directly
et al., 2011). In another study, the antiallergic agent Azelastine into matrix depositing myofibroblasts (Radisky, 2005; Andersson-
was administered in the food of mice that received an acute dose Sjöland et al., 2011). The process of EMT is mediated by Snail and
of radiation and reduced the severity of radiation dermatitis. In Twist transcription factors which repress E-Cadherin and promote
these animals there was a modest reduction in the severity of der- a mesenchymal phenotype enabling the epithelial cells to escape
matitis. The authors proposed that the mechanism responsible attachment from the basement membrane permitting anchor-
was increased epithelial cell stabilization post-radiation exposure age independent growth and migration into the interstitial space
due to preventing the influx of Ca2+ into the cell resulting in a (Willis et al., 2005); ultimately, the damaged alveoli collapse and
decrease in free radical generation (Murakami et al., 1997). Until are fully encompassed by connective tissue (Almeida et al., 2013).
an effective therapeutic is approved for this indication, patients The process of fibrosis occurs several months later than pneu-
will continue to be treated with standard burn agents such as monitis, typically appearing 6 months after radiation exposure.
Aloe Vera which neither prevents fibrosis nor addresses the pal- The timing of these late effects are supported by data show-
liative component associated with skin injury due to radiation ing that increased TGFβ signaling is sustained after pneumonitis
exposure. resolves thus promoting the latent fibrosis (Rube et al., 2000). Sim-
The clinical observation of heterogeneity in the manifesta- ilar signaling events have been described for bleomycin-induced
tion of fibrosis-related to radiation-induced dermatitis suggests pulmonary fibrosis. The common clinical practice of administer-
a possible genetic basis for risk. The results of a recent study ing concomitant chemoradiation (in this case, chemotherapy is
by Andreassen et al. (2013) in which cultured fibroblasts from administered as a radiosensitizer) increases the risk of severe and
head and neck cancer patients with subcutaneous fibrosis were extended pulmonary fibrosis. Mouse models of thoracic radiation
irradiated. Analysis of the cells provided validation of a group of and treatment with bleomycin have revealed molecular mecha-
overexpressed genes associated with a positive fibrosis phenotype. nisms driving the general induction process of fibrosis in the lung.
Additional studies on the predictive ability of synergistically acting In addition to the previously described classical TGFβ and TNF-α
genes, both in tissue and in peripheral blood, would certainly be signaling pathways that are responsible for initiating inflammation
of great interest. at the site of injury, there is subsequent involvement of chemokines
and their receptors in orchestrating the recruitment and trafficking
PULMONARY FIBROSIS of the immune cells from the circulation to the sites of inflam-
Radiation therapy is a critical component in the treatment of many mation driving the epithelial cell responses within the alveoli
forms of cancers of the lung, breast, and lymphomas. In these cases, (Johnston et al., 2002; Han et al., 2011). Interestingly, chemokine

Frontiers in Pharmacology | Inflammation Pharmacology March 2014 | Volume 5 | Article 51 | 6


Mancini and Sonis Cancer regimen-induced fibrosis

production differs significantly between the pneumonitic and (SODs) are naturally occurring enzymes in the body which act
fibrotic phases of injury suggesting differential roles for subsets of in the early stages of injury. SODs dismutate superoxides to
chemokine family members and likely specificity in the immune form hydrogen peroxide (H2 O2 )and catalase further decomposes
cells they recruit (Johnston et al., 1998). Whereas chronic inflam- H2 O2 to oxygen and water. Because radiation therapy reduces the
mation is a hallmark of fibrosis, sustained chemokine recruitment endogenous SOD, SOD mimetics have been developed for ther-
and trafficking of immune cells may lie at the crux of the cellular apeutic use and have demonstrated success in reducing fibrosis
events propagating the long term complications associated with by enhancing the activity of antioxidant enzymes in the tissue,
the condition. preventing oxidative damage with fewer side effects and higher
Like the majority of regimen-related toxicities, the risk of pul- potency than amifostine (Gao et al., 2012; Pan et al., 2012).
monary fibrosis is not ubiquitous among patients undergoing
treatment. In reality, despite demographic and treatment inten- CONCLUDING STATEMENTS
sity equivalence, only a relatively small fraction of patients go on Chronic problems associated with fibrosis are of high impor-
to develop fibrosis. This clinical observation has led to signifi- tance because they can ultimately force physicians to limit an
cant interest to determine those factors that are associated with individuals’ cancer treatment due to a decreased tolerance of the
risk. It now appears that genetic differences among patients, espe- therapies employed. As a result, there is a high demand to develop
cially those genes that are associated with the pathways leading interventions designed to prevent or treat fibrosis associated with
to fibrosis are fundamental determinants of risk. This hypothesis regimen-related toxicities. Animal models designed to mimic radi-
was elegantly demonstrated by Paun and Haston (2012) who com- ation and chemotherapy-induced fibrosis in patients and to help
pared radiation-induced pulmonary fibrosis susceptibility across understand the exact mechanisms of these pathways have revealed
a number of mouse syngeneic mouse strains and then, using a that there are marked differences in the cytokine responses among
genome-wide association study format, identified specific SNP loci animal strains. This is consistent with a varied cytokine response
associated with the condition. The complexity of the pathobiology seen in patients; therefore highly valuable information has been
of radiation-induced fibrosis and its impact on clinical risk predic- gained from collecting and incorporating data from several differ-
tion has been further substantiated by candidate SNP approaches. ent models. The discovery that many toxicities seem to cluster
As noted above, TGFβ1 has been associated with the biological suggests shared pathoetiologies that will not only assist in the
sequence leading to fibrosis. Consequently, TGFβ1 SNPs seemingly development of such therapeutics, but will also likely result in
made a good target for risk determination. Results of substantive the use of one therapy for the treatment or prevention of fibrosis
clinical studies have been, however, inconsistent (Barnett et al., in multiple tissue types.
2012). It seems likely that effective genomic risk prediction of
fibrosis will rely on the discovery of a cluster of SNPs or genes
REFERENCES
(Petrovski et al., 2009). Abdollani, A., Griggs, D. W., Zieher, H., Roth, A., Lipson, K. E., Saffrich, R., et al.
Preventive strategies such as the use of intensity-modulated (2005). Inhibition of alpha (v) beta 3 integrin survival signaling enhances antian-
radiation therapy (IMRT) have been suggested to reduce the giogenic and antitumor effects of radiotherapy. Clin. Cancer Res. 11, 6270–6279.
frequency and severity of radiation induced pulmonary fibro- doi: 10.1158/1078-0432.CCR-04-1223
sis (Lavrenkov et al., 2007). On the other hand, modifying Almeida, C., Nagarajan, D., Tian, J., Leal, S. W., Wheeler, K., Munley, M., et al.
(2013). The role ofPLoS ONE alveolar epithelium in radiation-induced lung
treatment-related strategies associated with chemotherapy is more injury. 8:e53628. doi: 10.1371/journal.pone.0053628
challenging given the wide range of drugs that are associated with Andersson-Sjöland, A., Nihlberg, K., Eriksson, L., Bjermer, L., and Westergren-
lung injury (Carver et al., 2007). Despite attempts to mitigate Thorsson, G. (2011). Fibrocytes and the tissue niche in lung repair. Respir. Res.
cancer therapy induced lung injury, pulmonary fibrosis persists 12, 76. doi: 10.1186/1465-9921-12-76
Andreassen, C. N., Overgaard, J., and Alsner, J. (2013). Independent prospective
and remains a significant side effect that is refractory to most
validation of a predictive test for risk of radiation induced fibrosis on the gene
treatments. Presently a lung transplant is viewed as the treat- expression pattern in fibroblasts irradiated in vitro. Radiother. Oncol. 108, 469–
ment of choice (Thabut et al., 2003) however, potential biologically 472. doi: 10.1016/j.radonc.2013.08.029
directed targets for intervention are emerging. For example, the Ara, G., Watkins, B. A., Zhong, H., Hawthorne, T. R., Karkaria, C. E., Sonis, S. T.,
Foxm1 transcription factor has been implicated in driving inflam- et al. (2008). Valifermin (rhFGF-20) reduces the severity and duration of hamster
cheek pouch mucositis induced by fractionated radiation. Int. J. Radiat. Biol. 84,
mation, EMT and proliferation of fibroblasts in the lung and
401–412. doi: 10.1080/09553000802007601
therefore might be of interest as a potential target for intervention Ashcroft, G., Yang, X., Glick, A. B., Weinstein, M., Letterio, J. L., Mizel, D.
(Balli et al., 2013). Additional treatment strategies for pulmonary E., et al. (1999). Mice lacking Smad3 show accelerated wound healing and an
fibrosis are similar to the previously described therapies for fibrosis impaired local inflammatory response. Nat. Cell Biol. 1, 260–266. doi: 10.1038/
from other tissue origins including agents which act as free rad- 12971
Balli, D., Ustiyan, V., Zhang, Y., Wang, I. C., Masino, A. J., Ren, X.,
ical scavengers. One such example approved by the FDA for use et al. (2013). Foxm1 transcription factor is required for lung fibrosis and
as a broad spectrum radioprotective agent is the thiol amifostine epithelial-to-mesenchymal transition. EMBO J. 32, 231–244. doi: 10.1038/emboj.
(Kouvaris et al., 2007). Clinical trial data suggests that amifostine 2012.336
protects against the long term effects of radiation and chemora- Barnett, G. C., Elliott, R. M., Alsner, J., Andreassen, C. N., Abdelhay, O., Burnet,
diation induced toxicity to lung and soft tissue including fibrosis. N. G., et al. (2012). Individual patient data meta-analysis shows no association
between the SNP rs1800469 in TGFB and late radiotherapy toxicity. Radiother.
Treatment with amifostine also allows for higher doses of radiation Oncol. 105, 289–295. doi: 10.1016/j.radonc.2012.10.017
to be administered in situations where that is required for higher Bey, E., Prat, M., Duhamel, P. D., Benderitter, M., Brachet, M., Trompier, F., et al.
tumor kill (Koukourakis et al., 2013). Superoxide dismutases (2010). Emerging therapy for improving wound repair of sever radiation burns

www.frontiersin.org March 2014 | Volume 5 | Article 51 | 7


Mancini and Sonis Cancer regimen-induced fibrosis

using local bone marrow-derived stem cell administrations. Wound Repair Regen. Haboubi, N. Y., Schofield, P. F., and Rowland, P. L. (1988). The light and electron
18, 50–58. doi: 10.1111/j.1524-475X.2009.00562.x microscopic features of early and late phase radiation-induced proctitis. Am. J.
Blirando, K., Milliat, F., Martelly, I., Sabourin, J. C., Benderitter, M., and François, Gastroenterol. 83, 1140–1144.
A. (2011). Mast cells are an essential component of human radiation proctitis Han, G., Zhang, H., Xie, C. H., and Zhou, Y. F. (2011). Th2-like immune
and contribute to experimental colorectal damage in mice. Am. J. Pathol. 178, response in radiation-induced lung fibrosis. Oncol. Rep. 26, 383–388. doi:
640–651. doi: 10.1016/j.ajpath.2010.10.003 10.3892/or.2011.1300
Boerma, M., Wang, J., Sridharan, V., Herbert, J. M., and Hauer-Jensen, M. (2013). Haselton, P. S., Carr, N., and Schofield, P. F. (1985). Vascular changes in
Pharmacological induction of transforming growth factor-beta1 in rat models radiation bowel disease. Histopathology 9, 517–534. doi: 10.1111/j.1365-
enhances radiation injury in the intestine and heart. PLoS ONE 8:e70479. doi: 2559.1985.tb02833.x
10.1371/journal.pone.0070479 Holmes, A., Abraham, D., Sa, S., Shiwen, X., Black, C. M., and Leask, A. (2001).
Brown, K., and Rzucidlo, E. (2011). Acute and chronic radiation injury. J. Vasc. Surg. CTGF and SMADs, maintenance of scleroderma phenotype is independent of
53, 15S–21S. doi:10.1016/j.jvs.2010.06.175 SMAD signaling. J. Biol. Chem. 276, 10594–10601. doi: 10.1074/jbc.M010149200
Brunn, T., and Fletcher, C. D. (2006). Postradiation vascular proliferations: Hugh, R. P., and Pemberton, A. D. (2002). Tissue-specific expression of mast cell
an increasing problem. Histophathology 48, 106–114. doi: 10.1111/j.1365- granule serine proteinases and their role in inflammation in the lung and gut.
2559.2005.02293.x Immunol. 105, 373–390. doi: 10.1046/j.1365-2567.2002.01375.x
Burdelya, L. G., Gleiberman, A. S., Toshkov, I., Aygun-Sunar, S., Bapardekar, M., Johnston, C. J., Williams, J. P., Okunieff, P., and Finkelstein, J. N.
Manderscheid-Kern, P., et al. (2011). Toll-like receptor 5 agonist protects mice (2002). Radiation-induced pulmonary fibrosis: examination of chemokine and
from dermatitis and oral mucositis caused by local radiation: implications for chemokine receptor families. Radiat. Res. 157, 256–265. doi: 10.1667/0033-
head-and-neck cancer radiotherapy. Int. J. Radiat. Oncol. Biol. Phys. 83, 228–234. 7587(2002)157[0256:RIPFEO]2.0.CO;2
doi: 10.1016/j.ijrobp.2011.05.055 Johnston, C. J., Wright, T. W., Rubin, P., and Finkelstein, J. N. (1998). Alter-
Carver, J. R., Shapiro, C. L., Ng, A., Jacobs, L., Schwartz, C., Virgo, K. S., et al. (2007). ations in the expression of chemokine mRNA levels in fibrosis-resistant and
American society of clinical oncology clinical evidence review on the ongoing -sensitive mice after thoracic radiation. Exp. Lung Res. 24, 321–337. doi:
care of adult cancer survivors: cardiac and pulmonary late effects. J. Clin. Oncol. 10.3109/01902149809041538
25, 3991–4008. doi: 10.1200/JCO.2007.10.9777 Kang, S., Chun, M., Jin, Y. M., Cho, M. S., Oh, Y. T., Ahn, B. O., et al. (2000). A rat
Caughey, G. H. (2007). Mast cell typtases and chymases in inflammation and host model for radiation-induced proctitis. J. Korean Med. Sci. 15, 682–689.
defense. Immunol. Rev. 217, 141–154. doi: 10.1111/j.1600-065X.2007.00509.x Koukourakis, M. I., Panteliadou, M., Abatzoglou, I. M., Sismanidou, K., Sivridis, E.,
Chan, R. J., Larsen, E., and Chan, P. (2012). Re-examining the evidence in radi- and Giatromanolaki, A. (2013). Postmastectomy hypofractionated and acceler-
ation dermatitis management literature: an overview and a critical appraisal ated radiation therapy with and without subcutaneous amifostine cytoprotection.
of systematic reviews. Int. J. Radiat. Oncol. Biol. Phys. 84, e357–e362. doi: Int. J. Radiat. Oncol. Biol. Phys. 85, e7–e13. doi: 10.1016/j.ijrobp.2012.08.017
10.1016/j.ijrobp.2012.05.009 Kouvaris, J., Kouloulias, V., and Vlahos, L. (2007). Amifostine: the first
Chen, E., Greenlee, B., Wills-Karp, M., and Moller, D. R. (2001). Attenua- selective-target and broad-spectrum radioprotector. Oncologist 12, 738–747. doi:
tion of lung inflammation and fibrosis in interferon–γ–deficient mice after 10.1634/theoncologist.12-6-738
intratracheal bleomycin. Am. J. Respir. Cell Mol. Biol. 24, 545–555. doi: Lataillade, J. J., Doucet, C., Bey, E., Carsin, H., Huet, C., Clairand, I., et al. (2007).
10.1165/ajrcmb.24.5.4064 New approach to radiation burn treatment by dosimetry-guided surgery com-
Counter, S. F., Froese, D. P., and Hart, M. J. (1999). Prospective evaluation of bined with autologous mesenchymal stem cell therapy. Regen. Med. 2, 785–794.
formalin therapy for radiation proctitis. Am. J. Surg. 177, 396–398. doi: 10.2217/17460751.2.5.785
Eckes, B., Zigrino, P., Kessler, D., Holtkötter, O., Shephard, P., Mauch, C., et al. Lavrenkov, K., Christian, J. A., Partridge, M., Niotsikou, E., Cook, G., Parker, M.,
(2000). Fibroblast-matrix interactions in wound healing and fibrosis. Matrix et al. (2007). A potential to reduce pulmonary toxicity: the use of perfusion
Biol. 19, 325–332. doi: 10.1016/S0945-053X(00)00077-9 SPECT with IMRT for functional lung avoidance in radiotherapy of non-small
Elting, L. S., Keefe, D. M., Sonis, S. T.,Garden, A. S., Spijkervet, F. K., Barasch, A., et al. cell lung cancer. Radiother. Oncol. 83, 156–162. doi: 10.1016/j.radonc.2007.
(2008). Patient-reported measurements of oral mucositis in head and neck cancer 04.005
patients treated with radiotherapy with or without chemotherapy: demonstration Leask, A., and Abraham, D. (2004). TGFβ Signaling and the fibrotic response. FASEB
of increased frequency, severity, resistance to palliation, and impact on quality of J. 18, 816–827. doi: 10.1096/fj.03-1273rev
life. Cancer 113, 2704–2713. doi: 10.1002/cncr.23898 Leask, A., Holmes, A., Black, C. M., and Abraham, D. J. (2003). Connective tissue
Fajardo, L. F. (2005). The pathology of ionizing radiation as defined by morphologic growth factor gene regulation. requirements for its induction by transform-
patterns. Acta Oncol. 44, 13–22. doi: 10.1080/02841860510007440 ing growth factor-beta 2 in fibroblasts. J. Biol. Chem. 278, 13008–13015. doi:
Flanders, K. C., Major, C. D., Arabshahi, A., Aburime, E. E., Okada, M. H., Fujii, M., 10.1074/jbc.M210366200
et al. (2003). Interference with transforming growth factor-b/Smad3 signaling Leiper, K., and Morris, A. I. (2007). Treatment of radiation proctitis. Clin. Oncol.
results in accelerated healing of wounds in previously irradiated skin. Am. J. 19, 724–729. doi: 10.1016/j.clon.2007.07.008
Pathol. 163, 2247–2257. doi: 10.1016/S0002-9440(10)63582-1 Linard, C., Busson, E., Holler, V., Strup-Perrot, C., Lacave-Lapalun, J. V., Lhomme,
Flanders, K. C., Sullivan, C. D., Fujii, M., Sowers, A., Anzano, M. A., Arabshahi, A., B., et al. (2013). Repeated autologous bone marrow-derived mesenchymal stem
et al. (2002). Mice lacking Smad3 are protected against cutaneous injury induced cell injections improve radiation-induced proctitis in pigs. Stem Cells Transl. Med.
by ionizing radiation. Am. J. Pathol. 160, 1057–1068. 2, 916–927. doi: 10.5966/sctm.2013-0030
Fujita-Yoshigaki, J., and Qi, B. (2009). Parotid acinar cells transiently change to duct- Lyons, A., Crichton, S., and Pezier, T. (2013). Trismus following radiotherapy to the
like cells during epithelial-mesenchymal transition. J. Med. Invest. 56, 258–259. head and neck is likely to have distinct genotype dependent cause. Oral Oncol. 49,
doi: 10.2152/jmi.56.258 932–936.
Gao, F., Fish, B. L., Szabo, A., Doctrow, S. R., Kma, L., Molthen, R. C., et al. (2012). Martin, M., Lefaix, J., and Delanian, S. (2000). TGF-β1 and radiation fibrosis: a
Short-term treatment with a SOD/catalase mimetic, EUK-207, mitigates pneu- master switch and a specific therapeutic target? Int. J. Radiat. Oncol. Biol. Phys.
monitis and fibrosis after single-dose total-body or whole-thoracic irradiation. 47, 277–290. doi: 10.1016/S0360-3016(00)00435-1
Radiat. Res. Soc. 178, 468–480. doi: 10.1667/RR2953.2 Massague, J. (2012). TGFβ signaling in context. Nat. Rev. Mol. Cell Biol. 13, 616–630.
Ghosh, A., Wei, J., Wu, M., and Varga J. (2008). Constitutive Smad signaling and doi: 10.1038/nrm3434
Smad-dependent collagen gene expression in mouse embryonic fibroblasts lack- Mehta, V. (2005). Radiation pneumonitis and pulmonary fibrosis in non-small cell
ing peroxisome proliferator-activated receptor–γ. Biochem. Biophys. Res. Comm. lung cancer: pulmonary function, prediction and prevention. Int. J. Radiat. Oncol.
374, 231–236. doi: 10.1016/j.bbrc.2008.07.014 Biol. Phys. 63, 5–24. doi: 10.1016/j.ijrobp.2005.03.047
Gurujeyalakshmi, G., and Giri, S. N. (1995). Molecular mechanisms of antifibrotic Mori, R., Kondo, T., Ohshima, T., Ishida, Y., and Mukaida, N. (2002). Accel-
effect of interferon gamma in bleomycin-mouse model of lung fibrosis: down- erated wound healing in tumor necrosis factor receptor p55-deficient mice
regulation of TGF-beta and procollagen I and III gene expression. Exp. Lung Res. with reduced leukocyte infiltration. FASEB 16, 963–974. doi: 10.1096/fj.01-
21, 791–808. doi: 10.3109/01902149509050842 0776com

Frontiers in Pharmacology | Inflammation Pharmacology March 2014 | Volume 5 | Article 51 | 8


Mancini and Sonis Cancer regimen-induced fibrosis

Murakami, R., Baba, Y., Nishimura, R., Furusawa, M., Yokoyama, T., Yamashita, Y., Sonis, S. T., and Keefe, D. M. (2013). The Pathobiology of Cancer
et al. (1997). The effect of azelastine on acute radiation dermatitis in mice mod- Regimen-Related Toxicities. New York: Springer. doi: 10.1007/978-1-4614-
els. Int. J. Radiat. Oncol. Biol. Phys. 37, 907–911. doi: 10.1016/S0360-3016(96) 5438-0
00621-9 Sonis, S. T., Peterson, R. L., Edwards, L. J., Lucey, C. A., Wang, L., Mason, L.,
Murray, L., Kramer, M., Hesson, D. P., Watkins, B. A., Fey, E. G., Argen- et al. (2000). Defining mechanisms of action of interleukin-11 on the progression
tieri, R. L., et al. (2010). Serum amyloid P ameliorates radiation-induced oral of radiation-induced oral mucositis in hamsters. Oral Oncol. 36, 373–381. doi:
mucositis and fibrosis. Fibrogenesis Tissue Repair 3, 11. doi: 10.1186/1755-1536- 10.1016/S1368-8375(00)00012-9
3-11 Sonnylal, S., Xu, S., Jones, H., Tam, A., Sreeram, V. R., Ponticos, M.,
Nathan, C. F., Prendergast, T. J., Wiebe, M. E., Stanley, E. R., Platzer, E., et al. (2013). Connective tissue growth factor causes EMT-like cell fate
Remold, H. G., et al. (1984). Activation of Human Macrophages. Compari- changes in vivo and in vitro. J. Cell Sci. 126, 2164-2175. doi: 10.1242/jcs.
son of other cytokines with interferon-gamma. J. Exp. Med. 160, 600–605. doi: 111302
10.1084/jem.160.2.600 Symon, Z., Goldshmidt, Y., Picard, O., Yavzori, M., Ben-Horin, S., Alezra, D., et al.
Oh, C. J., Kim, J. Y., Choi, Y. K., Kim, H. J., Jeong, J. Y., Bae, K. H., et al. (2012). (2010). A murine model for the study of molecular pathogenesis of pathogen-
Dimethylfumarate attenuates renal fibrosis via NF-E2-Related factor 2-Mediated esis of radiation proctitis. Int. J. Radiat. Oncol. Biol. Phys. 76, 242–250. doi:
inhibition of transforming growth factor-β/Smad signaling. PLoS ONE 7:e45870. 10.1016/j.ijrobp.2009.07.1736
doi: 10.1371/journal.pone.0045870 Thabut, G., Mal, H., Castier, Y., Groussard, O., Brugière, O., Marrash-
Okunieff, P., Cornelison, T., Mester, M., Liu, W., Ding, I., Chen, Y., et al. (2005). Chahla, R., et al. (2003). Survival benefit of lung transplantation for patients
Mechanism and modification of gastrointestinal soft tissue response to radia- with pulmonary fibrosis. J. Thorac. Cardiovasc. Surg. 126, 469–475. doi:
tion: role of growth factors. Int. J. Radiat. Oncol. Biol. Phys. 62, 273–278. doi: 10.1016/S0022-5223(03)00600-7
10.1016/j.ijrobp.2005.01.034 Tsoutsou, P., and Koukourakis, M. (2006). Radiation pneumonitis and fibrosis:
Pan, J., Su, Y., Hou, X., He, H., Liu, S., Wu, J., et al. (2012). Protective effect of mechanisms underlying its pathogenesis and implications for future research.
recombinant protein SOD-TAT on radiation – induced injury in mice. Life Sci. Int. J. Radiat. Oncol. Biol. Phys. 66, 1281–1293. doi: 10.1016/j.ijrobp.2006.
91, 89–93. doi: 10.1016/j.lfs.2012.06.003 08.058
Paun, A., and Haston, C. K. (2012). Genomic and genome-wide association of Ueha, S., Shand, F., and Matsushima, K. (2012). Cellular and molecular mechanism
susceptibility to radiation-induced fibrotic lung disease in mice. Radiother. Oncol. of chronic inflammation-associated organ fibrosis. Front. Immunol. 3:71. doi:
105, 350–357. doi: 10.1016/j.radonc.2012.08.004 10.3389/fimmu.2012.00071
Petrovski, S., Szoeke, C. E., Sheffield, L. J., D’souza, W., Huggins, R. M., and Venkov, C., Link, A., Jennings, J. L., Plieth, D., Inoue, T., Nagai, K., et al. (2007). A
O’brien, T. J. (2009). Multi-SNP pharmacogenomic classifier is superior to single- proximal activator of transcription in epithelial-mesenchymal transition. J. Clin.
SNP models for predicting drug outcome in complex diseases. Pharmacogenet. Invest. 117, 482–491. doi: 10.1172/JCI29544
Genomics 19, 147–152. doi: 10.1097/FPC.0b013e32831d1dfa Vermorken, J. B., and Specenier, P. (2010). Optimal treatment for recur-
Piguet, P. F., Collart, M. A., Grau, G. E., Sappino, A. P., and Vassalli, P. rent/metastatic head and neck cancer. Ann. Oncol. 21(Suppl. 7), vii252–vii261.
(1990). Requirement of tumour necrosis factor for development of silica-induced doi: 10.1093/annonc/mdq453
pulmonary fibrosis. Nature 344, 245–247. doi: 10.1038/344245a0 Wang, Q., Usinger, W., Nichols, B., Gray, J., Xu, L., Seeley, T. W., et al. (2011).
Radisky, D. (2005). Epithelial-mesenchyman transition. J.Cell Sci. 118, 4325–4326. Cooperative interaction of CTGF and TGF-b in animal models of fibrotic disease.
doi: 10.1242/jcs.02552 Fibrogenesis Tissue Repair 4:4. doi: 10.1186/1755-1536-4-4
Rosai, J. (1996). “Acute pulmonary injury and interstitial pneumonia,” in Ack- Willis, B. C., Liebler, J. M., Luby-Phelps, K., Nicholson, A. G., Crandall, E. D.,
erman’s Surgical Pathology, 8th Edn, Vol. 1, ed. J Rosai (New York: Mosby), du Bois, R. M., et al. (2005). Induction of epithelial-mesenchymal transition in
358–359. alveolar epithelial cells by transforming growth factor-beta1: a potential role in
Rube, C., Uthe, D., Schmid, K. W., Richter, K. D., Wessel, J., Schuck, A., et al. (2000). idiopathic pulmonary fibrosis. Am. J. Pathol. 166, 1321–1332. doi: 10.1016/S0002-
Dose-dependent induction of transforming growth factor β (TGFβ) in the lung 9440(10)62351-6
tissue of fibrosis-prone mice after thoracic irradiation. Int. J. Radiat. Oncol. Biol. Wong, R. K., Bensadoun, R., Boers-Doets, C. B., Bryce, J., Chan, A., Epstein,
Phys. 47, 1033–1042. doi: 10.1016/S0360-3016(00)00482-X J. B., et al. (2013). Clinical practice guidelines for the prevention and treat-
Ryan, J. (2012). Ionizing radiation: the good, the bad, and the ugly. J. Invest. ment of acute and late radiation reactions from the MASCC Skin Toxicity
Dermatol. 132, 985–993. doi: 10.1038/jid.2011.411 Study Group. Support Care Cancer 21, 2933–2948. doi: 10.1007/s00520-013-
Salvo, N., Barnes, E., van Draanen, J., Stacey, E., Mitera, G., Breen, D., et al. 1896-2
(2010). Prophylaxis and management of acute radiation-induced skin reactions: Zhu, M. L., Wang, M., Shi, T. Y., Li, Q. X., Xi, P., Xia, K. Q., et al.
a systematic review of the literature. Curr. Oncol. 17, 94–112. (2013). No association between TGFβ1 polymorphisms and late radiother-
Sempowski, G., Borrello, M., Blieden, T. M., Barth, R. K., and Phipps, R. P. (1995). apy toxicity: a meta-analysis. PLoS ONE 8:e76964. doi: 10.1371/journal.pone.
Fibroblast heterogeneity in the healing wound. Wound Repair Regen. 3, 120–131. 0076964
doi: 10.1046/j.1524-475X.1995.30204.x
Sheth, S., Bleibel, W., Thukral, C., A-Rahim, Y., Beldi, G., Csizmadia, E., et al. (2009). Conflict of Interest Statement: The authors declare that the research was conducted
Heightened NTPDase-1/CD39 expression and angiogenesis in radiation proctitis. in the absence of any commercial or financial relationships that could be construed
Purinergic Signal. 5, 321–326. doi: 10.1007/s11302-009-9154-2 as a potential conflict of interest.
Silverman, S. Jr. (2007). Diagnosis and management of oral mucositis. J. Support.
Oncol. 5(Suppl. 1), 13–21. Received: 04 November 2013; paper pending published: 13 February 2014; accepted:
Skwarchuk, M., and Travis, E. (1998). Changes in histology and fibrogenic cytokines 09 March 2014; published online: 27 March 2014.
in irradiated colorectum of two murine strains. Int. J. Radiat. Oncol. Biol. Phys. Citation: Mancini ML and Sonis ST (2014) Mechanisms of cellular fibrosis associ-
42, 169–178. doi: 10.1016/S0360-3016(98)00201-6 ated with cancer regimen-related toxicities. Front. Pharmacol. 5:51. doi: 10.3389/
Son, H.-N., Nam, J.-O., Kim, S., and Kim, I. S. (2013). Multiple FAS1 domains fphar.2014.00051
and the RGD motif of TGFBI act cooperatively to bind αvβ3 integrin, leading This article was submitted to Inflammation Pharmacology, a section of the journal
to anti-angiogenic and anti-tumor effects. Biochim. Biophys. Acta Mol. Cell Res. Frontiers in Pharmacology.
1833, 2378–2388. doi: 10.1016/j.bbamcr.2013.06.012 Copyright © 2014 Mancini and Sonis. This is an open-access article distributed under
Sonis, S. T. (2007). Pathobiology of mucositis: novel insights and opportunities. J. the terms of the Creative Commons Attribution License (CC BY). The use, distribution
Support. Oncol. 5, 3–11. or reproduction in other forums is permitted, provided the original author(s) or licensor
Sonis, S. T. (2011). Oral mucositis in head and neck cancer. Am. Soc. Clin. Oncol. are credited and that the original publication in this journal is cited, in accordance with
Educ. Book 236–240. accepted academic practice. No use, distribution or reproduction is permitted which
Sonis, S. T. (2013). Oral mucositis. Anticancer Drugs 47, 831–839. does not comply with these terms.

www.frontiersin.org March 2014 | Volume 5 | Article 51 | 9

Das könnte Ihnen auch gefallen